Review article
A20: A multifunctional tool for regulating immunity and preventing disease

https://doi.org/10.1016/j.cellimm.2019.04.002Get rights and content

Highlights

  • A20 is a potent anti-inflammatory protein that is strongly linked to multiple human diseases.

  • A20 deficiency in distinct cell types confers susceptibility to different disease phenotypes.

  • A20 restricts NF-κB signaling as well as other ubiquitin dependent signals.

  • A20 possesses several distinct biochemical motifs that regulate signals.

Abstract

A20, also known as TNFAIP3, is a potent regulator of ubiquitin (Ub) dependent signals. A20 prevents multiple human diseases, indicating that the critical functions of this protein are clinically as well as biologically impactful. As revealed by mouse models, cell specific functions of A20 are linked to its ability to regulate diverse signaling pathways. Aberrant expression or functions of A20 in specific cell types underlie divergent disease outcomes. Discernment of A20’s biochemical functions and their phenotypic outcomes will contribute to our understanding of how ubiquitination is regulated, how Ub mediated functions can prevent disease, and will pave the way for future therapeutic interventions.

Section snippets

A20 and the Ub mediated regulation of cellular processes

Protein ubiquitination is a post-translational modification that orchestrates diverse cellular functions. The prevalence of intracellular ubiquitination events is expansive, and proteomic analyses reveal a large fraction of the mammalian genome is dedicated to encoding Ub associated and Ub interacting proteins [1], [2], [3]. Substrate proteins may be modified by the addition of a single Ub (monoubiquitination) by an enzyme complex that includes an E1 Ub activating enzyme, an E2 Ub conjugating

A20’s links to human disease

A20, or TNFAIP3, is a remarkably potent regulator of Ub dependent signals and of immune homeostasis. Among ubiquitination regulators, A20 has garnered increasing attention because of its multiple links to human disease [15], [16]. Initially, genome wide association studies (GWAS) linked single nucleotide polymorphisms (SNPs) in the A20 gene locus with the incidence of rheumatoid arthritis, systemic lupus erythematosus (SLE), psoriasis, rheumatoid arthritis, celiac disease, inflammatory bowel

A20 and adaptive immunity

Most cell types express baseline levels of A20 protein that is further induced by stimulation with a variety of ligands such as TNF that trigger NF-κB signaling. The induction of A20 expression by pro-inflammatory signals suggests A20 is a negative feedback regulator of inflammation. Spontaneous activation of multiple immune cell types was first observed in globally A20 deficient (A20−/−) mice, leading to multi-organ inflammation and perinatal death [46]. A20’s broad expression presented

A20 functions in innate immunity

Spontaneous inflammation, perinatal lethality, and enhanced NF-κB signaling are the most prominent phenotypes in A20−/− mice. These inflammatory phenotypes persist in A20−/− RAG-1−/− compound mutant mice, indicating that A20 performs critical physiological functions independently of adaptive lymphocytes [46]. T and B cell independent regulatory functions of A20 may broadly be partitioned into regulation of innate immune cells or non-hematopoietic cells. Potent inhibition of innate immune

Non-hematopoietic functions of A20

A20 expression is induced in non-hematopoietic cells as well as immune cells. The genetic association of A20 with a variety of distinct autoimmune diseases—including diseases afflicting specific tissues—prompted investigations of A20’s physiological functions in tissue restricted cell types such as intestinal, lung, and skin epithelial cells. A20 deficiency in villin-expressing intestinal epithelial cells (IECs) renders mice susceptible to dextran sulfate sodium (DSS) [69]. This defect also

A20 beyond NF-κB signaling

Lineage specific studies of A20 have not only highlighted diverse cell type specific functions for this protein, but have also expanded A20’s cell autonomous functions. While A20’s capacity to prevent inflammation has largely been ascribed to its ability to restrict NF-κB signaling, A20 also appears to regulate additional Ub dependent signals.

While cells with deficient NF-κB signals are more susceptible to programmed cell death, A20 deficient fibroblasts and T cells are more susceptible to cell

Biochemical mechanisms by which A20’s mediates its functions

While A20 clearly regulates ubiquitination of multiple signaling complexes, the precise biochemical mechanisms by which A20 performs these functions remain surprisingly enigmatic and complex. The A20 protein is a cysteine protease with de-ubiquitinating (DUB) activity [45], [57]. A20 also possesses seven zinc finger motifs that mediate binding to at least two types of polyubiqutin chains, RIP1, IKKγ, TRAF2, and a number of additional Ub dependent proteins (e.g., ABIN-1, RNF11, TAX1BP1). Hence,

Outstanding questions/future areas for discovery

If A20 performs critical non-catalytic functions, then A20 may partly function as an adaptor and/or scaffold protein. This function could involve some of A20’s binding partners, several of which display overlapping signaling functions with A20. For example, ABIN-1 and TAX1BP1 share A20’s ability to restrict TNF and TLR induced NF-κB responses as well as TNF induced cell death [81], [82], [83], [84], [85]. A20 would be an unusual adaptor protein, as it is dynamically regulated at transcriptional

Acknowledgements

We apologize to contributors in this field that we have not had room to reference.

Work from the authors’ laboratory was supported by NIH R01 grants AI117908, DK095693, and AI35198.

References (88)

  • L. Catrysse et al.

    A20 in inflammation and autoimmunity

    Trends Immunol.

    (2014)
  • R. Arsenescu et al.

    Signature biomarkers in Crohn’s disease: toward a molecular classification

    Mucosal Immunol.

    (2008)
  • Q. Zhou et al.

    Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease

    Nat. Genet.

    (2016)
  • C. Franco-Jarava et al.

    TNFAIP3 haploinsufficiency is the cause of autoinflammatory manifestations in a patient with a deletion of 13Mb on chromosome 6

    Clin. Immunol.

    (2018)
  • F. Berteau et al.

    Autosomic dominant familial Behçet disease and haploinsufficiency A20: a review of the literature

    Autoimmun. Rev.

    (2018)
  • U. Novak et al.

    The NF-κB negative regulator TNFAIP3 (A20) is inactivated by somatic mutations and genomic deletions in marginal zone lymphomas

    Blood

    (2009)
  • G. Nocturne et al.

    Germline and somatic genetic variations of TNFAIP3 in lymphoma complicating primary Sjögren’s syndrome

    Blood

    (2013)
  • T. Ghadban et al.

    Evaluation of the germline single nucleotide polymorphism rs583522 in the TNFAIP3 gene as a prognostic marker in esophageal cancer

    Cancer Genet.

    (2015)
  • S.C. Pitt et al.

    Identification of novel oncogenic mutations in thyroid cancer

    J. Am. Coll. Surg.

    (2016)
  • A.W. Opipari et al.

    The A20 zinc finger protein protects cells from tumor necrosis factor cytotoxicity

    J. Biol. Chem.

    (1992)
  • R.M. Tavares et al.

    The ubiquitin modifying enzyme A20 restricts B cell survival and prevents autoimmunity

    Immunity

    (2010)
  • Y. Chu et al.

    B cells lacking the tumor suppressor TNFAIP3/A20 display impaired differentiation and hyperactivation and cause inflammation and autoimmunity in aged mice

    Blood

    (2011)
  • O. Hitotsumatsu et al.

    The ubiquitin-editing enzyme A20 restricts nucleotide-binding oligomerization domain containing 2-triggered signals

    Immunity

    (2008)
  • M. Kool et al.

    The ubiquitin-editing protein A20 prevents dendritic cell activation, recognition of apoptotic cells, and systemic autoimmunity

    Immunity

    (2011)
  • M. Valdearcos et al.

    Microglial inflammatory signaling orchestrates the hypothalamic immune response to dietary excess and mediates obesity susceptibility

    Cell Metab.

    (2017)
  • C. Schneider et al.

    A metabolite-triggered tuft cell-ILC2 circuit drives small intestinal remodeling

    Cell

    (2018)
  • M.A. O’Donnell et al.

    Ubiquitination of RIP1 regulates an NF-κB-independent cell-death switch in TNF signaling

    Curr. Biol.

    (2007)
  • B.H. Duong et al.

    A20 restricts ubiquitination of pro-interleukin-1β protein complexes and suppresses NLRP3 inflammasome activity

    Immunity

    (2015)
  • T.T. Lu et al.

    Dimerization and ubiquitin mediated recruitment of A20, a complex deubiquitinating enzyme

    Immunity

    (2013)
  • M.G. Kattah et al.

    Ubiquitin-modifying enzymes and regulation of the inflammasome

    J. Mol. Biol.

    (2017)
  • K. Heyninck et al.

    Structure–function analysis of the A20-binding inhibitor of NF-κB activation, ABIN-1

    FEBS Lett.

    (2003)
  • P. Beaudette et al.

    Proteomic techniques to probe the ubiquitin landscape

    Proteomics

    (2016)
  • R. Yau et al.

    The increasing complexity of the ubiquitin code

    Nat. Cell Biol.

    (2016)
  • E. Oh et al.

    Principles of ubiquitin-dependent signaling

    Ann. Rev. Cell Dev. Biol.

    (2018)
  • J.J. Sims et al.

    Polyubiquitin-sensor proteins reveal localization and linkage-type dependence of cellular ubiquitin signaling

    Nat. Methods

    (2012)
  • P. Cohen et al.

    The role of hybrid ubiquitin chains in the MyD88 and other innate immune signalling pathways

    Cell Death Differ.

    (2017)
  • S. Wang et al.

    An enhancer element harboring variants associated with systemic lupus erythematosus engages the TNFAIP3 promoter to influence A20 expression

    PLoS Genet.

    (2013)
  • I. Adrianto et al.

    Association of a functional variant downstream of TNFAIP3 with systemic lupus erythematosus

    Nat. Genet.

    (2011)
  • S. Wang et al.

    TALEN-mediated enhancer knockout influences TNFAIP3 gene expression and mimics a molecular phenotype associated with systemic lupus erythematosus

    Genes Immun.

    (2016)
  • U.K. Sokhi et al.

    Dissection and function of autoimmunity-associated TNFAIP3 (A20) gene enhancers in humanized mouse models

    Nat. Commun.

    (2018)
  • X. Jiang et al.

    Expression of tumor necrosis factor alpha-induced protein 3 mRNA in peripheral blood mononuclear cells negatively correlates with disease severity in psoriasis vulgaris

    Clin. Vaccine Immunol.

    (2012)
  • M.J. Schuijs et al.

    Farm dust and endotoxin protect against allergy through A20 induction in lung epithelial cells

    Science

    (2015)
  • A. Aki et al.

    Hypomorphic A20 expression confers susceptibility to psoriasis

    PLoS ONE

    (2017)
  • M. Devos et al.

    Keratinocyte expression of A20/TNFAIP3 controls skin inflammation associated with atopic dermatitis and psoriasis

    J. Invest. Dermatol.

    (2018)
  • Cited by (0)

    View full text